1887

Abstract

Coronavirus disease 2019 (COVID-19) identified in December 2019 in Wuhan, China, is associated with high mortality rates worldwide.

Thrombotic problems, such as coagulopathy, are common in COVID-19 patients. Despite anticoagulation, thrombosis is more common in patients in the intensive care unit and patients with more severe disease. Although the exact mechanisms of coagulopathy in COVID-19 patients are still unclear, studies showed that overactivation of the renin-angiotensin system (RAS), cytokine storm, endothelial damage, formation of neutrophil extracellular traps (NETs), and also extracellular vesicles (EVs) in response to COVID-19 induced inflammation can lead to systemic coagulation and thrombosis.

The management of COVID-19 patients requires the use of basic and readily available laboratory markers, both on admission and during hospitalization. Because it is critical to understand the pathophysiology of COVID-19 induced coagulopathy and treatment strategies, in this review we attempt to explain the underlying mechanism of COVID-19 coagulopathy, its diagnosis, and the associated successful treatment strategies.

The exact mechanisms behind COVID-19-related coagulopathy are still unclear, but several studies revealed some mechanisms. More research is needed to determine the best anticoagulant regimen and to study other therapeutic options.

  • This is an open-access article distributed under the terms of the Creative Commons Attribution NonCommercial License. The Microbiology Society waived the open access fees for this article.
Loading

Article metrics loading...

/content/journal/jmm/10.1099/jmm.0.001591
2022-11-08
2024-04-26
Loading full text...

Full text loading...

/deliver/fulltext/jmm/71/11/jmm001591.html?itemId=/content/journal/jmm/10.1099/jmm.0.001591&mimeType=html&fmt=ahah

References

  1. Kutti-Sridharan G, Vegunta R, Vegunta R, Mohan BP, Rokkam VRP. SARS-CoV2 in different body fluids, risks of transmission, and preventing COVID-19: a comprehensive evidence-based eview. Int J Prev Med 2020; 11:97 [View Article]
    [Google Scholar]
  2. Meyerowitz EA, Richterman A, Gandhi RT, Sax PE. Transmission of SARS-CoV-2: a review of viral, host, and environmental factors. Ann Intern Med 2021; 174:69–79 [View Article]
    [Google Scholar]
  3. Badawi S, Ali BR. ACE2 nascence, trafficking, and SARS-CoV-2 pathogenesis: the saga continues. Hum Genomics 2021; 15:1–14 [View Article]
    [Google Scholar]
  4. Sathler PC. Hemostatic abnormalities in COVID-19: A guided review. An Acad Bras Cienc 2020; 92:e20200834 [View Article]
    [Google Scholar]
  5. Bahraini M, Dorgalaleh A. The impact of SARS-CoV-2 infection on blood coagulation and fibrinolytic pathways: a review of prothrombotic changes caused by COVID-19. Semin Thromb Hemost 2022; 48:19–30 [View Article]
    [Google Scholar]
  6. Klok FA, Kruip MJHA, van der Meer NJM, Arbous MS, Gommers DAMPJ et al. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb Res 2020; 191:145–147 [View Article]
    [Google Scholar]
  7. McFadyen JD, Stevens H, Peter K. The emerging threat of (Micro)thrombosis in COVID-19 and Its therapeutic implications. Circ Res 2020; 127:571–587 [View Article]
    [Google Scholar]
  8. Qiu H, Yuan L, Huang X, Zhou Y, Wu Q et al. Using the big data of internet to understand the characteristics of coronavirus disease 2019: a big data study. Otolaryngol Head Neck Surg 2020569–575
    [Google Scholar]
  9. Miesbach W. Pathological role of angiotensin II in severe COVID-19. TH Open 2020; 4:e138–e144 [View Article]
    [Google Scholar]
  10. Jose RJ, Manuel A. COVID-19 cytokine storm: the interplay between inflammation and coagulation. Lancet Respir Med 2020; 8:e46–e47 [View Article]
    [Google Scholar]
  11. Gonzalez Cañas E, Gimenez Gaibar A, Rodriguez Lorenzo L, Castro Rios J, Martinez Toiran A et al. Acute peripheral arterial thrombosis in COVID-19. BRIT J SURG 2020; 107:e444–e445
    [Google Scholar]
  12. Zuo Y, Yalavarthi S, Shi H, Gockman K, Zuo M et al. Neutrophil extracellular traps (NETs) as markers of disease severity in COVID-19. medRxiv 2020; 2020:2020 [View Article]
    [Google Scholar]
  13. Rosell A, Havervall S, von Meijenfeldt F, Hisada Y, Aguilera K et al. Patients With COVID-19 have elevated levels of circulating extracellular vesicle tissue factor activity that is associated with severity and mortality-brief report. Arterioscler Thromb Vasc Biol 2021; 41:878–882 [View Article]
    [Google Scholar]
  14. Pena ALB, Oliveira RA, Severo RG, Simões E Silva AC. COVID-19 related coagulopathy: what is known up to now. Curr Med Chem 2021; 28:4207–4225 [View Article]
    [Google Scholar]
  15. Andersen KG, Rambaut A, Lipkin WI, Holmes EC, Garry RF. The proximal origin of SARS-CoV-2. Nat Med 2020; 26:450–452 [View Article]
    [Google Scholar]
  16. Marchandot B, Sattler L, Jesel L, Matsushita K, Schini-Kerth V et al. COVID-19 related coagulopathy: a distinct entity?. J Clin Med 2020; 9:E1651 [View Article]
    [Google Scholar]
  17. Pawlowski C, Venkatakrishnan AJ, Kirkup C, Berner G, Puranik A et al. Enoxaparin is associated with lower rates of mortality than unfractionated eparin in hospitalized COVID-19 patients. EClinicalMedicine 2021; 33:100774 [View Article]
    [Google Scholar]
  18. Beigel JH, Tomashek KM, Dodd LE, Mehta AK, Zingman BS et al. Remdesivir for the treatment of Covid-19 - final report. N Engl J Med 2020; 383:1813–1826 [View Article]
    [Google Scholar]
  19. Shereen MA, Khan S, Kazmi A, Bashir N, Siddique R. COVID-19 infection: origin, transmission, and characteristics of human coronaviruses. J Adv Res 2020; 24:91–98 [View Article]
    [Google Scholar]
  20. Lee C, Choi WJ. Overview of COVID-19 inflammatory pathogenesis from the therapeutic perspective. Arch Pharm Res 2021; 44:99–116 [View Article]
    [Google Scholar]
  21. Komarova NL, Schang LM, Wodarz D. Patterns of the COVID-19 pandemic spread around the world: exponential versus power laws. J R Soc Interface 2020; 17:20200518 [View Article]
    [Google Scholar]
  22. García LF. Immune response, inflammation, and the clinical spectrum of COVID-19. Front Immunol 2020; 11:1441 [View Article]
    [Google Scholar]
  23. Webb BJ, Peltan ID, Jensen P, Hoda D, Hunter B et al. Clinical criteria for COVID-19-associated hyperinflammatory syndrome: a cohort study. Lancet Rheumatol 2020; 2:e754–e763 [View Article]
    [Google Scholar]
  24. Song J-W, Zhang C, Fan X, Meng F-P, Xu Z et al. Immunological and inflammatory profiles in mild and severe cases of COVID-19. Nat Commun 2020; 11:1–10 [View Article] [PubMed]
    [Google Scholar]
  25. Piazza G, Morrow DA. Diagnosis, management, and pathophysiology of arterial and venous thrombosis in COVID-19. JAMA 2020; 324:2548–2549 [View Article]
    [Google Scholar]
  26. Fei Y, Tang N, Liu H, Cao W. Coagulation dysfunction a hallmark in COVID-19. Arch Pathol Lab Med 2020; 144:1223–1229
    [Google Scholar]
  27. Taylor FB, Toh CH, Hoots WK, Wada H, Levi M et al. Towards definition, clinical and laboratory criteria, and a scoring system for disseminated intravascular coagulation. Thromb Haemost 2001; 86:1327–1330 [View Article]
    [Google Scholar]
  28. Tang N, Li D, Wang X, Sun Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J Thromb Haemost 2020; 18:844–847 [View Article]
    [Google Scholar]
  29. Del Nonno F, Colombo D, Nardacci R, Falasca L. Fatal pulmonary arterial thrombosis in a COVID-19 patient, with asymptomatic history, occurred after swab negativization. Thromb J 2021; 19:1–6 [View Article]
    [Google Scholar]
  30. Beckman M, Nyrén S, Kistner A. A case-report of widespread pulmonary embolism in a middle-aged male seven weeks after asymptomatic suspected COVID 19 infection. Thromb J 2020; 18:1–4 [View Article]
    [Google Scholar]
  31. Levi M, Thachil J, Iba T, Levy JH. Coagulation abnormalities and thrombosis in patients with COVID-19. Lancet Haematol 2020; 7:e438–e440 [View Article]
    [Google Scholar]
  32. Wang X, Sahu KK, Cerny J. Coagulopathy, endothelial dysfunction, thrombotic microangiopathy and complement activation: potential role of complement system inhibition in COVID-19. J Thromb Thrombolysis 2021; 51:657–662 [View Article]
    [Google Scholar]
  33. Savla SR, Prabhavalkar KS, Bhatt LK. Cytokine storm associated coagulation complications in COVID-19 patients: Pathogenesis and management. Expert Rev Anti Infect Ther 2021; 19:1397–1413 [View Article]
    [Google Scholar]
  34. Ghazi L, Drawz P. Advances in understanding the renin-angiotensin-aldosterone system (RAAS) in blood pressure control and recent pivotal trials of RAAS blockade in heart failure and diabetic nephropathy. F1000Res 2017; 6:F1000 Faculty Rev-297 [View Article]
    [Google Scholar]
  35. Balakumar P, Jagadeesh G. A century old renin-angiotensin system still grows with endless possibilities: AT1 receptor signaling cascades in cardiovascular physiopathology. Cell Signal 2014; 26:2147–2160 [View Article]
    [Google Scholar]
  36. Kamińska M, Mogielnicki A, Stankiewicz A, Kramkowski K, Domaniewski T et al. Angiotensin II VIA AT1 receptors acclerates arterail. J Physiol Pharmacol 2005; 56:571–585
    [Google Scholar]
  37. Ridker PM, Gaboury CL, Conlin PR, Seely EW, Williams GH et al. Stimulation of plasminogen activator inhibitor in vivo by infusion of angiotensin II. Evidence of a potential interaction between the renin-angiotensin system and fibrinolytic function. Circulation 1993; 87:1969–1973 [View Article]
    [Google Scholar]
  38. Ekholm M, Kahan T, Jörneskog G, Bröijersen A, Wallén NH. Angiotensin II infusion in man is proinflammatory but has no short-term effects on thrombin generation in vivo. Thromb Res 2009; 124:110–115 [View Article]
    [Google Scholar]
  39. Gheblawi M, Wang K, Viveiros A, Nguyen Q, Zhong J-C et al. Angiotensin-converting enzyme 2: SARS-CoV-2 receptor and regulator of the renin-angiotensin system: celebrating the 20th anniversary of the discovery of ACE2. Circ Res 2020; 126:1456–1474 [View Article]
    [Google Scholar]
  40. Kuba K, Imai Y, Penninger JM. Angiotensin-converting enzyme 2 in lung diseases. Curr Opin Pharmacol 2006; 6:271–276 [View Article]
    [Google Scholar]
  41. Arentz M, Yim E, Klaff L, Lokhandwala S, Riedo FX et al. Characteristics and outcomes of 21 critically Ill patients with COVID-19 in washington state. JAMA 2020; 323:1612–1614 [View Article]
    [Google Scholar]
  42. Wiese OJ, Allwood BW, Zemlin AE. COVID-19 and the renin-angiotensin system (RAS): a spark that sets the forest alight?. Med Hypotheses 2020; 144:110231 [View Article]
    [Google Scholar]
  43. Patel VB, Clarke N, Wang Z, Fan D, Parajuli N et al. Angiotensin II induced proteolytic cleavage of myocardial ACE2 is mediated by TACE/ADAM-17: a positive feedback mechanism in the RAS. J Mol Cell Cardiol 2014; 66:167–176 [View Article]
    [Google Scholar]
  44. Ragab D, Salah Eldin H, Taeimah M, Khattab R, Salem R. The COVID-19 cytokine storm; what we know so far. Front Immunol 2020; 11:1446 [View Article]
    [Google Scholar]
  45. Liu QQ, Cheng A, Wang Y, Li H, Hu L et al. Cytokines and their relationship with the severity and prognosis of coronavirus disease 2019 (COVID-19): a retrospective cohort study. BMJ Open 2020; 10:e041471 [View Article]
    [Google Scholar]
  46. Escher R, Breakey N, Lämmle B. Severe COVID-19 infection associated with endothelial activation. Thromb Res 2020; 190:62 [View Article]
    [Google Scholar]
  47. Tanaka T, Narazaki M, Kishimoto T. Immunotherapeutic implications of IL-6 blockade for cytokine storm. Immunotherapy 2016; 8:959–970 [View Article]
    [Google Scholar]
  48. Burzynski LC, Humphry M, Pyrillou K, Wiggins KA, Chan JNE et al. The coagulation and immune systems are directly linked through the activation of interleukin-1α by thrombin. Immunity 2019; 50:1033–1042 [View Article]
    [Google Scholar]
  49. Thornton P, McColl BW, Greenhalgh A, Denes A, Allan SM et al. Platelet interleukin-1alpha drives cerebrovascular inflammation. Blood 2010; 115:3632–3639 [View Article]
    [Google Scholar]
  50. Bester J, Matshailwe C, Pretorius E. Simultaneous presence of hypercoagulation and increased clot lysis time due to IL-1β, IL-6 and IL-8. Cytokine 2018; 110:237–242 [View Article]
    [Google Scholar]
  51. Brunn GJ, Saadi S, Platt JL. Constitutive repression of interleukin-1alpha in endothelial cells. Circ Res 2008; 102:823–830 [View Article] [PubMed]
    [Google Scholar]
  52. Tunjungputri RN, Li Y, de Groot PG, Dinarello CA, Smeekens SP et al. The inter-relationship of platelets with interleukin-1β-mediated inflammation in humans. Thromb Haemost 2018; 118:2112–2125 [View Article]
    [Google Scholar]
  53. Cavalli G, Colafrancesco S, Emmi G, Imazio M, Lopalco G et al. Interleukin 1α: a comprehensive review on the role of IL-1α in the pathogenesis and treatment of autoimmune and inflammatory diseases. Autoimmun Rev 2021; 20:102763 [View Article]
    [Google Scholar]
  54. Kozma GT, Mészáros T, Bakos T, Hennies M, Bencze D et al. Mini-factor H modulates complement-dependent IL-6 and IL-10 release in an immune cell culture (PBMC) model: potential benefits against cytokine storm. Front Immunol 2021; 12:642860 [View Article]
    [Google Scholar]
  55. Subramaniam S, Scharrer I. Procoagulant activity during viral infections. Front Biosci 2018; 23:1060–1081 [View Article]
    [Google Scholar]
  56. Chauhan AJ, Wiffen LJ, Brown TP. COVID-19: a collision of complement, coagulation and inflammatory pathways. J Thromb Haemost 2020; 18:2110–2117 [View Article]
    [Google Scholar]
  57. Markiewski MM, Nilsson B, Ekdahl KN, Mollnes TE, Lambris JD. Complement and coagulation: strangers or partners in crime?. Trends Immunol 2007; 28:184–192 [View Article]
    [Google Scholar]
  58. Oikonomopoulou K, Ricklin D, Ward PA, Lambris JD. Interactions between coagulation and complement—their role in inflammation. Seminars in immunopathology Springer; 2012
    [Google Scholar]
  59. Ritis K, Doumas M, Mastellos D, Micheli A, Giaglis S et al. A novel C5a receptor-tissue factor cross-talk in neutrophils links innate immunity to coagulation pathways. J Immunol 2006; 177:4794–4802 [View Article]
    [Google Scholar]
  60. Ding YQ, Bian XW. Analysis of coronavirus disease-19 (COVID-19) based on SARS autopsy. Zhonghua Bing Li Xue Za Zhi 2020; 49:291–293 [View Article]
    [Google Scholar]
  61. Tian S, Hu W, Niu L, Liu H, Xu H et al. Pulmonary pathology of early-phase 2019 novel coronavirus (COVID-19) pneumonia in two patients with lung cancer. J Thorac Oncol 2020; 15:700–704 [View Article]
    [Google Scholar]
  62. Perico L, Benigni A, Casiraghi F, Ng LFP, Renia L et al. Immunity, endothelial injury and complement-induced coagulopathy in COVID-19. Nat Rev Nephrol 2021; 17:46–64 [View Article]
    [Google Scholar]
  63. Helms J, Tacquard C, Severac F, Leonard-Lorant I, Ohana M et al. High risk of thrombosis in patients with severe SARS-CoV-2 infection: a multicenter prospective cohort study. Intensive Care Med 2020; 46:1089–1098 [View Article]
    [Google Scholar]
  64. Sardu C, Gambardella J, Morelli MB, Wang X, Marfella R et al. Hypertension, thrombosis, kidney failure, and diabetes: is COVID-19 an endothelial disease? a comprehensive evaluation of clinical and basic evidence. J Clin Med 2020; 9:E1417 [View Article]
    [Google Scholar]
  65. Bermejo-Martin JF, Almansa R, Torres A, González-Rivera M, Kelvin DJ. COVID-19 as a cardiovascular disease: the potential role of chronic endothelial dysfunction. Cardiovasc Res 2020; 116:e132–e133 [View Article]
    [Google Scholar]
  66. Olson JD. D-dimer: an overview of hemostasis and fibrinolysis, assays, and clinical applications. Adv Clin Chem 2015; 69:1–46 [View Article]
    [Google Scholar]
  67. Yau JW, Teoh H, Verma S. Endothelial cell control of thrombosis. BMC Cardiovasc Disord 2015; 15:1–11 [View Article]
    [Google Scholar]
  68. Pons S, Fodil S, Azoulay E, Zafrani L. The vascular endothelium: the cornerstone of organ dysfunction in severe SARS-CoV-2 infection. Crit Care 2020; 24:353–358 [View Article]
    [Google Scholar]
  69. Golshiri K, Ataei Ataabadi E, Portilla Fernandez EC, Jan Danser AH, Roks AJM. The importance of the nitric oxide-cGMP pathway in age-related cardiovascular disease: focus on phosphodiesterase-1 and soluble guanylate cyclase. Basic Clin Pharmacol Toxicol 2020; 127:67–80 [View Article]
    [Google Scholar]
  70. Teimury A, Khameneh MT, Khaledi EM. Major coagulation disorders and parameters in COVID-19 patients. Eur J Med Res 2022; 27:1–10 [View Article]
    [Google Scholar]
  71. Iba T, Warkentin TE, Thachil J, Levi M, Levy JH. Proposal of the definition for COVID-19-associated oagulopathy. J Clin Med 2021; 10:E191 [View Article]
    [Google Scholar]
  72. Federici AB. The factor VIII/von Willebrand factor complex: basic and clinical issues. Haematologica 2003; 88:EREP02
    [Google Scholar]
  73. Prabhakaran P, Ware LB, White KE, Cross MT, Matthay MA et al. Elevated levels of plasminogen activator inhibitor-1 in pulmonary edema fluid are associated with mortality in acute lung injury. Am J Physiol Lung Cell Mol Physiol 2003; 285:L20–8 [View Article]
    [Google Scholar]
  74. Loghmani H, Conway EM. Exploring traditional and nontraditional roles for thrombomodulin. Blood 2018; 132:148–158 [View Article]
    [Google Scholar]
  75. O’Sullivan JM, Gonagle DM, Ward SE, Preston RJS, O’Donnell JS. Endothelial cells orchestrate COVID-19 coagulopathy. Lancet Haematol 2020; 7:e553–e555 [View Article]
    [Google Scholar]
  76. Goshua G, Pine AB, Meizlish ML, Chang C-H, Zhang H et al. Endotheliopathy in COVID-19-associated coagulopathy: evidence from a single-centre, cross-sectional study. Lancet Haematol 2020; 7:e575–e582 [View Article]
    [Google Scholar]
  77. Macfarlane R, Fibrinolysis BR. Its mechanism and significance. Blood 1948; 3:1167–1187
    [Google Scholar]
  78. Moore HB, Moore EE. Temporal changes in fibrinolysis following injury. Semin Thromb Hemost 2020; 46:189–198 [View Article]
    [Google Scholar]
  79. Schmitt FCF, Manolov V, Morgenstern J, Fleming T, Heitmeier S et al. Acute fibrinolysis shutdown occurs early in septic shock and is associated with increased morbidity and mortality: results of an observational pilot study. Ann Intensive Care 2019; 9:1–15 [View Article]
    [Google Scholar]
  80. Sisson TH, Simon RH. The plasminogen activation system in lung disease. Curr Drug Targets 2007; 8:1016–1029 [View Article]
    [Google Scholar]
  81. Pavoni V, Gianesello L, Pazzi M, Stera C, Meconi T et al. Evaluation of coagulation function by rotation thromboelastometry in critically ill patients with severe COVID-19 pneumonia. J Thromb Thrombolysis 2020; 50:281–286 [View Article]
    [Google Scholar]
  82. Helms J, Tacquard C, Severac F, Leonard-Lorant I, Ohana M et al. High risk of thrombosis in patients with severe SARS-CoV-2 infection: a multicenter prospective cohort study. Intensive Care Med 2020; 46:1089–1098 [View Article]
    [Google Scholar]
  83. Wright FL, Vogler TO, Moore EE, Moore HB, Wohlauer MV et al. Fibrinolysis shutdown correlation with thromboembolic events in severe COVID-19 nfection. J Am Coll Surg 2020; 231:193–203 [View Article]
    [Google Scholar]
  84. Edler C, Schröder AS, Aepfelbacher M, Fitzek A, Heinemann A et al. Dying with SARS-CoV-2 infection-an autopsy study of the first consecutive 80 cases in hamburg, Germany. Int J Legal Med 2020; 134:1275–1284 [View Article]
    [Google Scholar]
  85. Meizoso JP, Moore HB, Moore EE. Fibrinolysis shutdown in COVID-19: clinical manifestations, molecular mechanisms, and therapeutic implications. J Am Coll Surg 2021; 232:995–1003 [View Article]
    [Google Scholar]
  86. Semeraro F, Ammollo CT, Morrissey JH, Dale GL, Friese P et al. Extracellular histones promote thrombin generation through platelet-dependent mechanisms: involvement of platelet TLR2 and TLR4. Blood 2011; 118:1952–1961 [View Article]
    [Google Scholar]
  87. Lagunas-Rangel FA. Neutrophil-to-lymphocyte ratio and lymphocyte-to-C-reactive protein ratio in patients with severe coronavirus disease 2019 (COVID-19): a meta-analysis. J Med Virol 2020; 92:1733–1734 [View Article]
    [Google Scholar]
  88. Iba T, Levy JH, Raj A, Warkentin TE. Advance in the management of sepsis-induced coagulopathy and disseminated intravascular coagulation. J Clin Med 2019; 8:E728 [View Article]
    [Google Scholar]
  89. Ali RA, Gandhi AA, Meng H, Yalavarthi S, Vreede AP et al. Adenosine receptor agonism protects against NETosis and thrombosis in antiphospholipid syndrome. Nat Commun 2019; 10:1–12 [View Article]
    [Google Scholar]
  90. Potey PM, Rossi AG, Lucas CD, Dorward DA. Neutrophils in the initiation and resolution of acute pulmonary inflammation: understanding biological function and therapeutic potential. J Pathol 2019; 247:672–685 [View Article]
    [Google Scholar]
  91. Meher AK, Spinosa M, Davis JP, Pope N, Laubach VE et al. Novel role of IL (interleukin)-1β in neutrophil extracellular trap formation and abdominal aortic aneurysms. Arterioscler Thromb Vasc Biol 2018; 38:843–853 [View Article]
    [Google Scholar]
  92. Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS et al. COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet 2020; 395:1033–1034 [View Article]
    [Google Scholar]
  93. Fuchs TA, Brill A, Wagner DD. Neutrophil extracellular trap (NET) impact on deep vein thrombosis. Arterioscler Thromb Vasc Biol 2012; 32:1777–1783 [View Article]
    [Google Scholar]
  94. Cedervall J, Zhang Y, Huang H, Zhang L, Femel J et al. Neutrophil extracellular traps accumulate in peripheral blood vessels and compromise organ function in tumor-bearing animals. Cancer Res 2015; 75:2653–2662 [View Article]
    [Google Scholar]
  95. Fuchs TA, Brill A, Duerschmied D, Schatzberg D, Monestier M et al. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci U S A 2010; 107:15880–15885 [View Article]
    [Google Scholar]
  96. Laridan E, Martinod K, De Meyer SF. Neutrophil Extracellular Traps in Arterial and Venous Thrombosis. Semin Thromb Hemost 2019; 45:86–93 [View Article]
    [Google Scholar]
  97. Bonow RO, Fonarow GC, O’Gara PT, Yancy CW. Association of coronavirus disease 2019 (COVID-19) with myocardial injury and mortality. JAMA Cardiol 2020; 5:751–753 [View Article]
    [Google Scholar]
  98. Meldolesi J. Extracellular vesicles, news about their role in immune cells: physiology, pathology and diseases. Clin Exp Immunol 2019; 196:318–327 [View Article]
    [Google Scholar]
  99. Słomka A, Urban SK, Lukacs-Kornek V, Żekanowska E, Kornek M. Large extracellular vesicles: have we found the holy grail of inflammation?. Front Immunol 2018; 9:2723 [View Article]
    [Google Scholar]
  100. Fogarty H, Townsend L, Ni Cheallaigh C, Bergin C, Martin-Loeches I et al. COVID19 coagulopathy in caucasian patients. Br J Haematol 2020; 189:1044–1049 [View Article]
    [Google Scholar]
  101. Terpos E, Ntanasis-Stathopoulos I, Elalamy I, Kastritis E, Sergentanis TN et al. Hematological findings and complications of COVID-19. Am J Hematol 2020; 95:834–847 [View Article]
    [Google Scholar]
  102. Balbi C, Burrello J, Bolis S, Lazzarini E, Biemmi V et al. Circulating extracellular vesicles are endowed with enhanced procoagulant activity in SARS-CoV-2 infection. EBioMedicine 2021; 67:103369 [View Article]
    [Google Scholar]
  103. Bautista-Vargas M, Bonilla-Abadía F, Cañas CA. Potential role for tissue factor in the pathogenesis of hypercoagulability associated with in COVID-19. J Thromb Thrombolysis 2020; 50:479–483 [View Article]
    [Google Scholar]
  104. Assimakopoulos SF, Emmanuil A, Tsimeka A, Chalkidi T, Marangos M et al. Evidence for increased circulating procoagulant phospholipids in patients with COVID-19 pneumonia and their prognostic role. Clin Chem Lab Med 2020; 59:e53–e55 [View Article]
    [Google Scholar]
  105. Zaid Y, Puhm F, Allaeys I, Naya A, Oudghiri M et al. Platelets Can Associate with SARS-Cov-2 RNA and Are Hyperactivated in COVID-19. Circ Res 2020 [View Article]
    [Google Scholar]
  106. Sinauridze EI, Kireev DA, Popenko NY, Pichugin AV, Panteleev MA et al. Platelet microparticle membranes have 50- to 100-fold higher specific procoagulant activity than activated platelets. Thromb Haemost 2007; 97:425–434 [PubMed]
    [Google Scholar]
  107. Cappellano G, Raineri D, Rolla R, Giordano M, Puricelli C et al. Circulating platelet-derived extracellular vesicles are a hallmark of Sars-Cov-2 infection. Cells 2021; 10:E85 [View Article]
    [Google Scholar]
  108. Lanyu Z, Feilong H. Emerging role of extracellular vesicles in lung injury and inflammation. Biomed Pharmacother 2019; 113:108748 [View Article]
    [Google Scholar]
  109. Wang J, Chen S, Bihl J. Exosome-mediated transfer of ACE2 (Angiotensin-Converting Enzyme 2) from Endothelial progenitor cells promotes survival and function of endothelial cell. Oxid Med Cell Longev 2020; 2020:4213541 [View Article]
    [Google Scholar]
  110. Kwon Y, Nukala SB, Srivastava S, Miyamoto H, Ismail NI et al. Exosomes facilitate transmission of SARS-cov-2 genome into human induced pluripotent stem cell-derived cardiomyocytes; 2020
  111. Harenberg J, Favaloro E. COVID-19: progression of disease and intravascular coagulation - present status and future perspectives. Clin Chem Lab Med 2020; 58:1029–1036 [View Article] [PubMed]
    [Google Scholar]
  112. Asakura H, Ogawa H. COVID-19-associated coagulopathy and disseminated intravascular coagulation. Int J Hematol 2021; 113:45–57 [View Article] [PubMed]
    [Google Scholar]
  113. Demelo-Rodríguez P, Cervilla-Muñoz E, Ordieres-Ortega L, Parra-Virto A, Toledano-Macías M et al. Incidence of asymptomatic deep vein thrombosis in patients with COVID-19 pneumonia and elevated D-dimer levels. Thromb Res 2020; 192:23–26 [View Article] [PubMed]
    [Google Scholar]
  114. Voicu S, Bonnin P, Stépanian A, Chousterman BG, Le Gall A et al. High prevalence of deep vein thrombosis in mechanically ventilated COVID-19 patients. J Am Coll Cardiol 2020; 76:480–482 [View Article]
    [Google Scholar]
  115. Eljilany I, Elzouki A-N. D-Dimer, Fibrinogen, and IL-6 in COVID-19 patients with suspected venous thromboembolism: a narrative review. Vasc Health Risk Manag 2020; 16:455–462 [View Article]
    [Google Scholar]
  116. Fu Y, Cheng Y, Wu Y. Understanding SARS-CoV-2-mediated inflammatory responses: from mechanisms to potential therapeutic tools. Virol Sin 2020; 35:266–271 [View Article]
    [Google Scholar]
  117. Zou Y, Guo H, Zhang Y, Zhang Z, Liu Y et al. Analysis of coagulation parameters in patients with COVID-19 in Shanghai, China. Biosci Trends 2020; 14:285–289 [View Article]
    [Google Scholar]
  118. Maier CL, Truong AD, Auld SC, Polly DM, Tanksley C-L et al. COVID-19-associated hyperviscosity: a link between inflammation and thrombophilia?. Lancet 2020; 395:1758–1759 [View Article] [PubMed]
    [Google Scholar]
  119. Ranucci M, Ballotta A, Di Dedda U, Baryshnikova E, Dei Poli M et al. The procoagulant pattern of patients with COVID-19 acute respiratory distress syndrome. J Thromb Haemost 2020; 18:1747–1751 [View Article]
    [Google Scholar]
  120. Thachil J, Agarwal S. Understanding the COVID-19 coagulopathy spectrum. J. Anesth 2020; 75:1432–1436 [View Article]
    [Google Scholar]
  121. Giannis D, Ziogas IA, Gianni P. Coagulation disorders in coronavirus infected patients: COVID-19, SARS-CoV-1, MERS-CoV and lessons from the past. J Clin Virol 2020; 127:104362 [View Article]
    [Google Scholar]
  122. Connors JM, Levy JH. Thromboinflammation and the hypercoagulability of COVID-19. J Thromb Haemost 2020; 18:1559–1561 [View Article]
    [Google Scholar]
  123. Thachil J, Tang N, Gando S, Falanga A, Cattaneo M et al. ISTH interim guidance on recognition and management of coagulopathy in COVID-19. J Thromb Haemost 2020; 18:1023–1026 [View Article]
    [Google Scholar]
  124. Jin X, Duan Y, Bao T, Gu J, Chen Y et al. The values of coagulation function in COVID-19 patients. PLoS ONE 2020; 15:e0241329 [View Article]
    [Google Scholar]
  125. Haimei MA. Pathogenesis and treatment strategies of COVID-19-related hypercoagulant and thrombotic complications. Clin Appl Thromb Hemost 2020; 26:1076029620944497 [View Article]
    [Google Scholar]
  126. Beigel J, Tomashek K, Dodd L. Remdesivir for the treatment of covid-19—final report. October 2020; 8:EJMoa2007764
    [Google Scholar]
  127. Food U, Administration D. FDA approves first treatment for COVID-19. Journal of food and drug analy 2020; 22:
    [Google Scholar]
  128. Tomazini BM, Maia IS, Cavalcanti AB, Berwanger O, Rosa RG et al. Effect of dexamethasone on days alive and ventilator-free in patients with moderate or severe acute respiratory distress syndrome and COVID-19: the CoDEX randomized clinical trial. JAMA 2020; 324:1307–1316 [View Article]
    [Google Scholar]
  129. Horby P, Lim WS, Emberson JR, Mafham M, Bell JL et al. Dexamethasone in hospitalized atients with Covid-19. N Engl J Med 2021; 384:693–704 [View Article]
    [Google Scholar]
  130. Coronavirus (COVID-19) update: FDA authorizes monoclonal antibodies for treatment of COVID-19. Journal of food and drug analy 2021
    [Google Scholar]
  131. Sujaritha J, Deepa sankar N, Mathivathani K, Aravindh G, Gnanasekaran G. An overview of some drugs: Lopinavir, Ritonavir, Chloroquine, Hydroxy chloroquine and Interferon as a effective treatment against COVID-19. ijrpp 2020; 10:20–24 [View Article]
    [Google Scholar]
  132. Lokugamage K, Hage A, Schindewolf C, Rajsbaum R, Menachery V. SARS-cov-2 sensitive to type I interferon pretreatment. bioRxiv 2020
    [Google Scholar]
  133. Hung IF-N, Lung K-C, Tso EY-K, Liu R, Chung TW-H et al. Triple combination of interferon beta-1b, lopinavir-ritonavir, and ribavirin in the treatment of patients admitted to hospital with COVID-19: an open-label, randomised, phase 2 trial. Lancet 2020; 395:1695–1704 [View Article]
    [Google Scholar]
  134. Stebbing J, Phelan A, Griffin I, Tucker C, Oechsle O et al. COVID-19: combining antiviral and anti-inflammatory treatments. Lancet Infect Dis 2020; 20:400–402 [View Article]
    [Google Scholar]
  135. Stebbing J, Krishnan V, de Bono S, Ottaviani S, Casalini G et al. Mechanism of baricitinib supports artificial intelligence-predicted testing in COVID-19 patients. EMBO Mol Med 2020; 12:e12697 [View Article]
    [Google Scholar]
  136. Sims JT, Krishnan V, Chang C-Y, Engle SM, Casalini G et al. Characterization of the cytokine storm reflects hyperinflammatory endothelial dysfunction in COVID-19. J Allergy Clin Immunol 2021; 147:107–111 [View Article]
    [Google Scholar]
  137. Cantini F, Niccoli L, Matarrese D, Nicastri E, Stobbione P et al. Baricitinib therapy in COVID-19: a pilot study on safety and clinical impact. J Infect 2020; 81:318–356 [View Article]
    [Google Scholar]
  138. Hoang TN, Pino M, Boddapati AK, Viox EG, Starke CE et al. Baricitinib treatment resolves lower-airway macrophage inflammation and neutrophil recruitment in SARS-CoV-2-infected rhesus macaques. Cell 2021; 184:460–475 [View Article]
    [Google Scholar]
  139. Kalil AC, Patterson TF, Mehta AK, Tomashek KM, Wolfe CR et al. Baricitinib plus remdesivir for hospitalized dults with Covid-19. N Engl J Med 2021; 384:795–807 [View Article]
    [Google Scholar]
  140. Martínez-Sanz J, Muriel A, Ron R, Herrera S, Pérez-Molina JA et al. Effects of tocilizumab on mortality in hospitalized patients with COVID-19: a multicentre cohort study. Clin Microbiol Infect 2021; 27:238–243 [View Article]
    [Google Scholar]
  141. Kewan T, Covut F, Al-Jaghbeer MJ, Rose L, Gopalakrishna KV et al. Tocilizumab for treatment of patients with severe COVID-19: A retrospective cohort study. EClinicalMedicine 2020; 24:100418 [View Article] [PubMed]
    [Google Scholar]
  142. Wei Q, Lin H, Wei R-G, Chen N, He F et al. Tocilizumab treatment for COVID-19 patients: a systematic review and meta-analysis. Infect Dis Poverty 2021; 10:71 [View Article]
    [Google Scholar]
  143. Raimondo MG, Biggioggero M, Crotti C, Becciolini A, Favalli EG. Profile of sarilumab and its potential in the treatment of rheumatoid arthritis. Drug Des Devel Ther 2017; 11:1593–1603 [View Article]
    [Google Scholar]
  144. Benucci M, Giannasi G, Cecchini P, Gobbi FL, Damiani A et al. COVID-19 pneumonia treated with Sarilumab: a clinical series of eight patients. J Med Virol 2020; 92:2368–2370 [View Article]
    [Google Scholar]
  145. Montesarchio V, Parrela R, Iommelli C, Bianco A, Manzillo E et al. Outcomes and biomarker analyses among patients with COVID-19 treated with interleukin 6 (IL-6) receptor antagonist sarilumab at a single institution in Italy. J Immunother Cancer 2020; 8:e001089 [View Article]
    [Google Scholar]
  146. Hodge JA, Kawabata TT, Krishnaswami S, Clark JD, Telliez J-B et al. The mechanism of action of tofacitinib - an oral Janus kinase inhibitor for the treatment of rheumatoid arthritis. Clin Exp Rheumatol 2016; 34:318–328
    [Google Scholar]
  147. Guimarães PO, Quirk D, Furtado RH, Maia LN, Saraiva JF et al. Tofacitinib in patients hospitalized with Covid-19 pneumonia. N Engl J Med 2021; 385:406–415 [View Article]
    [Google Scholar]
  148. Maslennikov R, Ivashkin V, Vasilieva E, Chipurik M, Semikova P et al. Tofacitinib reduces mortality in coronavirus disease 2019 Tofacitinib in COVID-19. Pulm Pharmacol Ther 2021; 69:102039 [View Article]
    [Google Scholar]
  149. Razonable RR, Pawlowski C, O’Horo JC, Arndt LL, Arndt R et al. Casirivimab-Imdevimab treatment is associated with reduced rates of hospitalization among high-risk patients with mild to moderate coronavirus disease-19. EClinicalMedicine 2021; 40:101102 [View Article]
    [Google Scholar]
  150. Lemos ACB, do Espírito Santo DA, Salvetti MC, Gilio RN, Agra LB et al. Therapeutic versus prophylactic anticoagulation for severe COVID-19: a randomized phase II clinical trial (HESACOVID). Thromb Res 2020; 196:359–366 [View Article]
    [Google Scholar]
  151. Lye DCB, Hui DS, Marks KM, Bruno R et al. Remdesivir for 5 or 10 days in patients with Severe Covid-19. N Engl J Med 2020; 383:1827–1837 [View Article]
    [Google Scholar]
  152. Kalil AC, Patterson TF, Mehta AK, Tomashek KM, Wolfe CR et al. Baricitinib plus remdesivir for hospitalized adults with Covid-19. N Engl J Med 2021; 384:795–807 [View Article]
    [Google Scholar]
  153. Spinner CD, Gottlieb RL, Criner GJ, Arribas López JR, Cattelan AM et al. Effect of remdesivir vs standard care on clinical status at 11 days in patients with moderate COVID-19: a randomized clinical trial. JAMA 2020; 324:1048–1057 [View Article]
    [Google Scholar]
  154. Tomazini BM, Maia IS, Bueno FR, Silva MVAO, Baldassare FP et al. COVID-19-associated ARDS treated with DEXamethasone (CoDEX): study design and rationale for a randomized trial. Rev Bras Ter Intensiva 2020; 32:354–362 [View Article]
    [Google Scholar]
  155. Rashad A, Mousa S, Nafady-Hego H, Nafady A, Elgendy H. Short term survival of critically ill COVID-19 Egyptian patients on assisted ventilation treated by either Dexamethasone or Tocilizumab. Sci Rep 2021; 11:8816 [View Article]
    [Google Scholar]
  156. Gottlieb RL, Nirula A, Chen P, Boscia J, Heller B et al. Effect of bamlanivimab as monotherapy or in combination with etesevimab on viral load in patients with mild to moderate COVID-19: a randomized clinical trial. JAMA 2021; 325:632–644 [View Article]
    [Google Scholar]
  157. Dougan M, Nirula A, Azizad M, Mocherla B, Gottlieb RL et al. Bamlanivimab plus etesevimab in mild or moderate COVID-19. N Engl J Med 2021; 385:1382–1392 [View Article]
    [Google Scholar]
  158. Davoudi-Monfared E, Rahmani H, Khalili H, Hajiabdolbaghi M, Salehi M et al. A randomized clinical trial of the efficacy and safety of interferon β-1a in treatment of severe COVID-19. Antimicrob Agents Chemother 2020; 64:e01061-20 [View Article]
    [Google Scholar]
  159. Rahmani H, Davoudi-Monfared E, Nourian A, Khalili H, Hajizadeh N et al. Interferon β-1b in treatment of severe COVID-19: a randomized clinical trial. Int Immunopharmacol 2020; 88:106903 [View Article]
    [Google Scholar]
  160. Bosi E, Bosi C, Rovere Querini P, Mancini N, Calori G et al. Interferon β-1a (IFNβ-1a) in COVID-19 patients (INTERCOP): study protocol for a randomized controlled trial. Trials 2020; 21:1–11 [View Article]
    [Google Scholar]
  161. Monk PD, Marsden RJ, Tear VJ, Brookes J, Batten TN et al. Safety and efficacy of inhaled nebulised interferon beta-1a (SNG001) for treatment of SARS-CoV-2 infection: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Respir Med 2021; 9:196–206 [View Article]
    [Google Scholar]
  162. Salama C, Han J, Yau L, Reiss WG, Kramer B et al. Tocilizumab in patients hospitalized with Covid-19 pneumonia. N Engl J Med 2021; 384:20–30 [View Article]
    [Google Scholar]
  163. Wang D, Fu B, Peng Z, Yang D, Han M et al. Tocilizumab in patients with moderate or severe COVID-19: a randomized, controlled, open-label, multicenter trial. Front Med 2021; 15:486–494 [View Article]
    [Google Scholar]
  164. Gordon AC, Angus DC, Derde LPG. Interleukin-6 receptor antagonists in critically Ill patients with Covid-19. reply. N Engl J Med 2021; 385:1147–1149 [View Article]
    [Google Scholar]
  165. Zhao H, Zhu Q, Zhang C, Li J, Wei M et al. Tocilizumab combined with favipiravir in the treatment of COVID-19: a multicenter trial in a small sample size. Biomed Pharmacother 2021; 133:110825 [View Article]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jmm/10.1099/jmm.0.001591
Loading
/content/journal/jmm/10.1099/jmm.0.001591
Loading

Data & Media loading...

This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error